Next Article in Journal
In Vitro Induction of Apoptosis in Isolated Acute Myeloid Leukemia Cells: The Role of Anastatica hierochuntica Methanolic Extract
Previous Article in Journal
Angiopoietin-Like 4 (ANGPTL4) in Patients with Psoriasis, Lichen Planus and Vitiligo—A Pilot Study from the Bialystok+ Polish Longitudinal University Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Hydrogen Sulfide Targeting Autophagy in the Pathological Processes of the Nervous System

1
Institute of Chronic Disease Risks Assessment, Henan University, Jinming Avenue, Kaifeng 475004, China
2
School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
*
Author to whom correspondence should be addressed.
Metabolites 2022, 12(9), 879; https://doi.org/10.3390/metabo12090879
Submission received: 11 August 2022 / Revised: 13 September 2022 / Accepted: 15 September 2022 / Published: 17 September 2022
(This article belongs to the Section Cell Metabolism)

Abstract

:
Autophagy is an important cellular process, involving the transportation of cytoplasmic contents in the double membrane vesicles to lysosomes for degradation. Autophagy disorder contributes to many diseases, such as immune dysfunction, cancers and nervous system diseases. Hydrogen sulfide (H2S) is a volatile and toxic gas with a rotten egg odor. For a long time, it was considered as an environmental pollution gas. In recent years, H2S is regarded as the third most important gas signal molecule after NO and CO. H2S has a variety of biological functions and can play an important role in a variety of physiological and pathological processes. Increasingly more evidences show that H2S can regulate autophagy to play a protective role in the nervous system, but the mechanism is not fully understood. In this review, we summarize the recent literatures on the role of H2S in the pathological process of the nervous system by regulating autophagy, and analyze the mechanism in detail, hoping to provide the reference for future related research.

1. Introduction

Autophagy refers to a complex molecular pathway in which intracellular components are transported to the lysosome chamber for degradation and recycling [1,2]. According to the different ways in which substrates enter lysosomes, there are three types of autophagy: macroautophagy, microautophagy and chaperone-mediated autophagy [3]. As an evolutionarily conservative process, autophagy helps cells adapt to various stress conditions by providing amino acid libraries through the decomposition of proteins and peptides. Therefore, autophagy maintains intracellular homeostasis, enabling cells to survive [4]. Autophagy dysfunction is associated with many diseases, such as cancer, metabolic diseases, neurodegenerative diseases and lung diseases [3,5].
Hydrogen sulfide (H2S) is a volatile, flammable and toxic gas with a rotten egg smell, which can be detected by the human nose at a very low content [6]. Recently, it has been regarded as a biological signal molecule together with nitric oxide (NO) and carbon monoxide (CO) [7,8]. H2S is involved in many physiological and pathological processes in the body, such as ischemia-reperfusion injury, vasodilation, carcinogenesis or inhibition of cancer, anti-inflammatory and regulation of hormone metabolism [9]. An increasing amount of evidences indicate that H2S regulates autophagy in many diseases, such as ischemia/reperfusion injury [10], lung disease [11] and neurodegenerative disease [12]. In this review, we summarize recent studies on the role of H2S in the pathological processes of the nervous system by regulating autophagy, and analyze the mechanism in detail, hoping to provide references for future related research.

2. Overview of Autophagy

Autophagy is a catabolic process through which cellular components, including proteins, lipids and organelles, are degraded in lysosomes and recycled to promote cellular homeostasis [13]. In the process of autophagy, the abnormal organelles and proteins, and pathogens are wrapped in autophagosomes formed by double membranes, and then transferred to lysosomes to be degraded [14]. Three types of autophagy have been found, including macroautophagy, microautophagy and chaperone-mediated autophagy, according to the substrate selectivity and the delivery path of the cargo to the lysosome cavity [15,16]. Macroautophagy, commonly known as autophagy, is the most thoroughly studied one, including initiation, expansion, closure and degradation processes [17]. Macroautophagy firstly forms cytosolic double membrane vesicles called autophagosomes to isolate the cargo. The autophagosomes then fuse with lysosomes to form autophagolysosomes, and the cargo is then degraded by the protease [18]. Chaperone-mediated autophagy transports a single unfolded and labeled protein directly across the lysosomal membrane. Microautophagy involves the direct uptake of cargo through lysosomal membrane invagination. All three types of autophagy lead to the degradation of cargo and transport the decomposition products back to the cytoplasm for cell reuse (Figure 1) [14,19,20,21,22,23,24,25]. Under physiological conditions, autophagy is usually at a basic level. Activated by various cellular stresses, including nutrient/energy starvation, endoplasmic reticulum stress, hypoxia, hypoxia, and organelle damage, the enhanced autophagy can clear the abnormal proteins in cells to maintain cell survival [26]. However, if autophagy is maintained at a high level for a long time due to internal and external factors, it may lead to autophagic death of cells to cause diseases. Therefore, the effect of autophagy on cells is a “double-edged sword” [27]. Autophagy disorders can be involved in a variety of pathological processes, including type 2 diabetes and obesity, infectious diseases and inflammation, neurodegenerative diseases and cancers [28]. In the pathological processes, the abnormal autophagy lost the function of clearing abnormal substances in the cell, leading to autophagic death [29]. However, the mechanism is not completely clear.

3. Overview of H2S

H2S is a colorless, flammable, lipophilic molecule with an unpleasant smell, similar to rotten eggs [30]. For many years, H2S has been simply regarded as a toxic gas and environmental pollutant [31]. Abe and Kimura’s 1996 report proposed the role of endogenous H2S in neural regulation, ushering in a new era of H2S study and its role in biology [32]. The evidence shows that H2S can regulate the function of the nervous system, especially the hippocampus. H2S not only plays an important regulatory role in the nervous system, but also plays an important role in digestive, cardiovascular system, urinary and blood systems. Therefore, H2S has been considered as the third gas signal molecule after CO and NO [33]. Endogenous H2S is mainly produced under the catalysis of three enzymes:cystathionine-β-synthase(CBS), cystathionine-γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST)[34]. During the production of endogenous H2S, L-homocysteine (Hcy) is converted into cystathionine, which is then converted into L-cysteine. CBS and CSE catalyse L-cysteine to produce H2S. L-cysteine was catalyzed by CAT to generate 3-mercaptopyruvate (3-MP) and eventually H2S. Moreover, 3-MST catalyzes 3-MP to produce H2S. Hcy is also catalyzed by CSE to produce H2S (Figure 2) [35,36]. H2S has many physiological functions, such as anti-apoptosis, anti-inflammatory, anti-oxidative stress, vasodilation and lowering blood pressure [37]. The biological function of H2S is mainly achieved by reversible protein vulcanization [38]. H2S mainly plays its physiological function by regulating cell function. There are many mechanisms for its regulation of cell function: histone modification, DNA methylation, DNA damage repair and H2S post-translational modification of proteins through sulfur hydration [39]. In recent years, the evidence has shown that H2S plays an important role by regulating autophagy in the pathological processes of the nervous system, including traumatic brain injury, nervous system hypoxia-ischemia injury, sleep deprivation-induced cognitive impairment, diabetic depression and Parkinson’s disease. However, the relevant mechanisms have not been fully understood. In this review, we summarize the literature on the role of H2S in regulating autophagy in the pathological processes of the nervous system, and analyze the related mechanism, in order to provide a reference for future research.

4. H2S Plays a Protective Role by Regulating Autophagy in Traumatic Brain Injury

Traumatic brain injury (TBI) refers to the interruption of brain function or other pathological changes of the brain caused by external forces. It is estimated that the annual incidence rate of TBI in the world is 50 million cases, and TBI is the major cause of the disability and death worldwide [40,41,42]. The secondary injury (subsequent biochemical changes) of TBI can lead to cell death, such as autophagic cell death and apoptosis, resulting in neurological impairment. Therefore, the inhibition of secondary cell death is the focus of brain injury treatment [43,44]. Mingyang Zhang and colleagues found that exogenous H2S ameliorated TBI of mice by decreasing brain edema, improving movement disorder and spatial memory acquisition after brain injury. Mechanism research revealed that H2S decreased the acute plasmalemma permeability in injured cells of the cortical and hippocampal brain regions in mice with TBI. The plasma membrane permeability is a marker of apoptosis and autophagy. Therefore, the subsequent detection of apoptosis and autophagy showed that H2S abolished TBI-induced cleaved caspase-3 and decline of Bcl-2, inhibited LC3-II, Beclin-1 and Vps34 activation and reversed the decline of p62 in the cortex and hippocampus of mice with TBI, indicating that H2S suppressed apoptosis and autophagy in TBI model of mice [45]. It has been reported that autophagic death and apoptosis participate in TBI [46]. Hence, in the above study, it can be deduced that exogenous H2S improves TBI of mice through inhibiting autophagy and apoptosis, which needs to be further confirmed [45]. In addition, Beclin 1 interacts with Bcl-2 through its BH3 domain [47], suggesting that autophagy and apoptosis can regulate each other. In the above, H2S reverses the upregulation of the Beclin-1/Bcl-2 ratio induced by TBI, which indicated that H2S inhibits apoptosis and autophagy through regulating Beclin-1-Vps34 interaction.
Mitochondria is a dynamic and multifunctional organelle, which plays an important role in maintaining the balance of intracellular environment and the function and survival of cells [48,49]. It has been reported that protecting mitochondria is important for TBI [50,51,52]. The results of Kebin Xu et al. showed that exogenous H2S preserved the integrity of blood–brain barrier (BBB) by increasing the expression of adherens junctions (AJs) and tight junctions (TJs), ameliorating pericyte survival, and mitigating neurovascular defect. H2S also protected neurons from apoptosis through decreasing apoptotic cells number and increasing Bcl-2/Bax ratio. Moreover, H2S induced remyelination and axonal repair through stabilizing microtubules and mitigating mitochondrial dysfunction. In addition, H2S suppressed autophagy following TBI, which was caused by the activation of the PI3K/AKT/mTOR pathway. Rapamycin (an autophagy activator) reversed H2S protection of TBI, while 3-MA (an autophagy suppressor) had the opposite effect, indicating that H2S improved TBI by inhibiting autophagy. Collectively, exogenous H2S ameliorated TBI through suppressing autophagy via activating PI3K/AKT/mTOR pathway [53]. The study showed that autophagy can promote cell survival through eliminating the damaged organelles and protein [27]. In H2S improvement of TBI, autophagy promotes cell death after TBI via the excessive degradation of basic cellular components, which is inhibited by H2S.
In addition to exogenous H2S, the endogenous H2S may also improve TBI. 3-MST is an important enzyme regulating endogenous H2S synthesis [54,55]. To explore 3-MST changes after TBI and its possible role, Mingyang Zhang et al. established a mouse model of TBI through a controlled cortical impingement system. The results showed that 3-MST existed in the cerebral cortex of normal mice. It increased gradually to reach a peak on the first day after TBI, and then dropped to a valley on the third day. Moreover, 3-MST collocated with neuron. Additionally, autophagy also peaked evidenced by the increased expression of LC3 on the first day after TBI. Moreover, the TBI-induced 3-MST was partially labeled by LC3. This indicated that some of the neurons expressing 3-MST, not dying neurons, were LC3 positive. However, 3-MST was not collocated with propidium iodide (cell death marker), and LC3 positive cells were partially colocalized with propidium iodide, suggesting that a considerable proportion of dead cells underwent autophagic cell death, and 3-MST has a protective effect on brain injury [56]. The evidence indicates that autophagy can maintain the survival of nerve cells [57,58]. Therefore, in the above study, it can be deduced that the TBI-induced 3-MST in cerebral cortex is related to the autophagic protection of neurons after TBI, suggesting that endogenous H2S may play an important role in autophagic cell death after TBI [56]. Whether endogenous H2S can improve TBI through autophagy needs further research.

5. H2S Plays a Protective Role by Regulating Autophagy in Nervous System Hypoxia-Ischemia Injury

5.1. H2S Plays a Protective Role by Regulating Autophagy in Spinal Cord Ischemia-Reperfusion Injury

Spinal cord ischemia-reperfusion (I/R) injury is a dynamic process and one of the most devastating complications during thoracic-abdominal aortic surgery, which can lead to the severe nerve defect of lower limbs and even brain death [59,60]. However, the pathological mechanism of spinal cord I/R injury is not completely clear, and there is no effective neuroprotective therapy [61]. It has been reported that autophagy is involved in spinal cord I/R injury; however, whether autophagy plays a protective or harmful role in spinal cord I/R injury is still uncertain [62,63]. Lei Li and colleagues established an in vivo and in vitro spinal cord I/R injury model and conducted a series of experiments. The results showed that exogenous H2S decreased the infarcted area of spinal cord and ameliorated the motor function of hind limbs of a rat model of spinal cord I/R injury. Mechanism research showed that H2S treatment decreased miR-30c expression and induced autophagy by upregulating the expression of Beclin-1 and LC3II in spinal cord of rat with spinal cord I/R injury. The results in OGD-induced spinal cord I/R injury of SY-SH-5Y cells were similar to those in vivo. Moreover, miR-30c negatively regulated Beclin-1 expression by targeting its 3′UTR, indicating that miR-30c negatively regulated autophagy in spinal cord with I/R injury. Similarly, exogenous H2S also suppressed Beclin-1 3′UTR in SY-SH-5Y cells with Oxygen, Glucose Deprivation (OGD)-induced spinal cord I/R injury. In addition, pretreatment of 3-MA or pre-miR-30c abolished H2S improvement of spinal cord I/R injury, indicating that exogenous H2S improved spinal cord I/R injury by promoting autophagy through inhibiting miR-30c [64]. The study indicates that in the case of extensive mitochondrial damage, autophagy can clear the damaged mitochondria to protect cells before it releases death-inducing proteins [63,65]. In the early stage of I/R injury, autophagy is upregulated to protect cells [66]. Therefore, in the above study, it can be deduced that H2S can improve spinal I/R injury by promoting autophagy and clearing the damaged mitochondria caused by I/R injury. Furthermore, it has been reported that the inhibition of autophagy improves spinal cord I/R injury [62], which is contrary to the above conclusion. The reason may be the different periods of spinal I/R injury, which needs to be studied further.

5.2. H2S Plays a Protective Role by Regulating Autoophagy in the Hypoxia-Ischemia Brain Injury of Neonatal Mice

Perinatal brain injury induced by hypoxia-ischemia (HI) may lead to neurodevelopmental disorders. Improving perinatal care can greatly improve the survival of infants with brain injury. In the critical period of brain development, HI can lead to perinatal brain neuron excitotoxicity, brain cell apoptosis and microglia activation [67,68]. H2S has been reported to play neuroprotective role in the central nervous system [69,70]. However, whether H2S can improve HI brain injury through regulating autophagy is not clear. Danqing Xin et al. found that L-Cysteine treatment after HI decreased early brain injury and improved behavioral deficits and synaptic damage in neonatal mice, which is related to the increased expression of synaptophysin and postsynaptic density protein 95 expression in the damaged cortex. In-depth research showed that L-cysteine could reduce the aggregation of CD11b+/CD45high cells, inhibit the activation of microglia and astrocytes, and decrease the upregulation of reactive oxygen species (ROS), malondialdehyde, neuronal apoptosis and inflammatory gene expression induced by HI in the damaged cortex of neonatal mice. Furthermore, L-Cysteine promoted autophagy by upregulating the expression of LC3 II and Beclin1 and downregulating p62 expression in the injured cortex after HI. CQ, an inhibitor of autophagy, abolished the protective effect of L-Cysteine on HI brain injury, indicating that L-Cysteine improved HI brain injury by promoting autophagy. In addition, the treatment of amino-oxyacetic acid (a suppressor of the H2S-producing enzyme) reversed the protective effect of L-Cysteine on HI brain injury. Collectively, endogenous H2S produced by L-Cysteine ameliorated HI-induced brain injury of neonatal mice by promoting autophagy [71]. In the above study, the enhanced autophagy can reduce ROS-mediated cell injury by scavenging HI-induced damaged mitochondria. Previous studies have shown that exogenous H2S promotes autophagy through the mTOR pathway [11,72,73]. Furthermore, Stat3 pathway has been reported to be involved in autophagy [74]. L-Cysteine inhibited mTOR and Stat3 pathway, suggesting that the endogenous H2S produced by L-Cysteine might promote autophagy by inhibiting the mTOR and Stat3 pathway, which needs to be further confirmed [71].

6. H2S Plays a Protective Role by Regulating Autophagy in Sleep Deprivation-Induced Cognitive Impairment

Sleep is very important to maintain the balance of physiological internal environment. Therefore, insufficient sleep will lead to various diseases. Sleep deprivation (SD) is an increasingly serious health problem in contemporary society [75]. SD can lead to cognitive impairment; however, the mechanism is not completely clear [76,77]. H2S has been reported to improve cognitive impairment [78]; however, how H2S inhibits SD-induced cognitive impairment has not been thoroughly studied. The results of Shan Gao and colleagues showed that exogenous H2S alleviated SD-induced cognitive impairment by ameliorating working memory impairment in Y-maze test, cognitive dysfunction in the novel object recognition test, location memory deficit in object location test, and spatial learning and memory disorder in the Morris water maze test, which were reversed by the inhibition of Sirt-1 by Sirtinol (an inhibitor of Sirit-1). H2S also reduced SD-induced hippocampal excessive autophagy by decreasing autophagosomes, downregulating Beclin1, and upregulating p62 in the hippocampus of SD-exposed rats. Furthermore, Sirtinol reversed H2S inhibition of the cognitive impairment and excessive hippocampal autophagy induced by SD in rats [79]. In addition, it has been reported that H2S increased Sirt-1 expression in the hippocampus of SD-exposed rats [80]. Collectively, it can be deduced that exogenous H2S mitigates SD-induced cognitive impairment by inhibiting autophagy via hippocampal Sirt-1 of rats [79]. Another study demonstrated that hippocampal excessive autophagy and inhibition of endogenous H2S production results in SD-induced cognitive impairment [81], which further confirmed the protective effect of H2S on SD-induced cognitive impairment.

7. H2S Plays a Protective Role by Regulating Autophagy in Diabetic Depression

The incidence rate of depression in people with diabetes is higher than that in people without diabetes [82,83]. Therefore, it is particularly important to study the mechanism of depression in diabetes. Brain-derived neurotropic factor (BDNF) has been reported to play an important role in depression [84,85]; however, the mechanism is not completely clear. Hai Yao Liu and colleagues found that exogenous H2S activated BDNF-TrkB pathway by increasing the protein expressions of BDNF and p-TrkB in the hippocampus of streptozotocin (STZ)-induced diabetic rats. K252a, which is an inhibitor of BDNF-TrkB pathway, abolished the antidepressant effect of H2S as evidenced by the tail suspension, novelty suppressed feeding, forced swimming and elevated plus-maze tests. Moreover, K252a reversed H2S-promoted hippocampal autophagy by downregulating the protein expression level of Beclin-1 and upregulating the protein expression of p62 in diabetic rats. Summarily, exogenous H2S improved depression by promoting autophagy via activating the BDNF-TrkB pathway [86]. In the above studies, in addition to BDNF-TrkB pathway, whether H2S can regulate autophagy through other ways to play an antidepressant role remains to be studied. In addition, the evidence indicates that the injury of hippocampal neurons contributes to diabetic depression [87,88]. Hence, future studies are needed to clarify whether H2S inhibits hippocampal neuronal damage by regulating autophagy.

8. H2S Plays a Protective Role by Regulating Autophagy in Parkinson’s Disease

Parkinson’s disease (PD) is a common progressive neurodegenerative disorder, the prevalence of which rises with advancing age. It affects about 2% of the population worldwide [89,90,91]. The neuronal apoptosis in the substantia has been reported to contribute to PD [92,93]. Wu Jiang et al. found that exogenous H2S mitigated neuronal apoptosis in the substantia by inhibiting 6-hydroxydopamine (OHDA)-induced TUNEL-positive cells, caspase-3 activity and Bax expression and mitigated 6-OHDA-induced reduction of Bcl-2 expression in substantia nigra of rats. In-depth research showed that 6-OHDA upregulated the expressions of Beclin-1, LC3-II and P62, increased the autophagosomes number and decreased the autolysosomes number in the substantia nigra, which were reversed by NaHS treatment, indicating that exogenous H2S restored the autophagy flux of substantia nigra impaired by 6-OHDA in rats. Moreover, H2S abolished 6-OHDA-induced decrease of leptin expression in the substantia nigra, and leptin-OBR, an inhibitor of leptin signaling, mitigated H2S inhibition of neuronal apoptosis and H2S promotion of the impaired autophagy in substantia nigra of rats treated by 6-OHDA. Summarily, exogenous H2S ameliorated neuronal apoptosis in substantia nigra by promoting autophagy impaired by 6-OHDA via activating leptin signaling in PD, which needed to be further confirmed by using the autophagy inhibitor [94]. It has been reported that mitochondrial dysfunction plays an important role in PD [95,96]. In the above study, exogenous H2S upregulates Bcl-2 expression and downregulates Bax expression, suggesting that H2S may inhibit mitochondrial-mediated neuronal apoptosis, which need to be studied further. Evidence indicates that the leptin signaling promotes autophagy [97,98]. The in-depth mechanism of H2S alleviating neuronal apoptosis through upregulating autophagy via leptin remains to be clarified.

9. Conclusions

In this review, we summarize the recent studies about the role of H2S targeting autophagy in the pathological processes of the nervous system as follows: (1) exogenous H2S ameliorates TBI of mice through suppressing autophagy and apoptosis; (2) exogenous H2S improves TBI by inhibiting autophagy via activating PI3K/AKT/mTOR pathway; (3) endogenous H2S may play protective role against TBI by inhibiting autophagic cell death; (4) exogenous H2S ameliorates spinal I/R injury through promoting autophagy and clearing the damaged mitochondria caused by I/R injury; (5) endogenous H2S produced by L-Cysteine improves HI-induced brain injury of neonatal mice by promoting autophagy via inhibiting mTOR and Stat3 pathway; (6) exogenous H2S alleviates SD-induced cognitive impairment by inhibiting autophagy via hippocampal Sirt-1; (7) exogenous H2S ameliorates depression through promoting autophagy by activating BDNF-TrkB pathway; (8) exogenous H2S improves neuronal apoptosis in substantia nigra through promoting autophagy impaired by 6-OHDA via activating leptin signaling (Table 1). It can be seen from the above that H2S sometimes promotes autophagy, and sometimes inhibits autophagy to protect the nervous system. The reason may be related to the types of nervous system diseases and the different course of nervous system disease. Generally speaking, at the beginning of the pathological process, the enhanced autophagy can help cells adapt to in vitro and in vivo stimulation and promote intracellular homeostasis, while the continuously enhanced autophagy can lead to autophagic death, thereby aggravating the pathological process. In addition, H2S regulates autophagy in the nervous system through a variety of pathways, including PI3K/AKT/mTOR pathway, mTOR/Stat3 pathway, Sirt-1 pathway, BDNF-TrkB pathway and leptin pathway. Whether there are other pathways to participate remains to be clarified in future research.
Our previous studies have shown that exogenous H2S can target autophagy/NLRP3 inflammasome and play a protective role in the liver [72,99]. Therefore, whether H2S can improve nervous system diseases by regulating autophagy/NLRP3 inflammasome is a topic worthy of study in the future.
With the deepening of relevant studies, H2S inhibition of autophagy death of neural cells may become a new therapeutic strategy for the treatment of neurological diseases.

Author Contributions

H.W. devised and co-wrote the manuscript; H.Z. co-wrote the manuscript and funded the manuscript; H.L. co-wrote the manuscript; Y.Y. drew the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare that there are no conflict of interest.

References

  1. Klionsky, D.J.; Petroni, G.; Amaravadi, R.K.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cadwell, K.; Cecconi, F.; Choi, A.M.K.; et al. Autophagy in major human diseases. EMBO J. 2021, 40, e108863. [Google Scholar] [CrossRef] [PubMed]
  2. Russo, M.; Russo, G.L. Autophagy inducers in cancer. Biochem. Pharmacol. 2018, 153, 51–61. [Google Scholar] [CrossRef] [PubMed]
  3. Liao, S.X.; Sun, P.P.; Gu, Y.H.; Rao, X.M.; Zhang, L.Y.; Ou-Yang, Y. Autophagy and pulmonary disease. Ther. Adv. Respir. Dis. 2019, 13, 1753466619890538. [Google Scholar] [CrossRef] [PubMed]
  4. Saha, S.; Panigrahi, D.P.; Patil, S.; Bhutia, S.K. Autophagy in health and disease: A comprehensive review. Biomed. Pharmacother. 2018, 104, 485–495. [Google Scholar] [CrossRef]
  5. Zhao, Y.G.; Zhang, H. Core autophagy genes and human diseases. Curr. Opin. Cell Biol. 2019, 61, 117–125. [Google Scholar] [CrossRef]
  6. Landry, A.P.; Ballou, D.P.; Banerjee, R. Hydrogen Sulfide Oxidation by Sulfide Quinone Oxidoreductase. Chembiochem 2021, 22, 949–960. [Google Scholar] [CrossRef]
  7. Roubenne, L.; Marthan, R.; Le Grand, B.; Guibert, C. Hydrogen Sulfide Metabolism and Pulmonary Hypertension. Cells 2021, 10, 1477. [Google Scholar] [CrossRef]
  8. Zhang, J.; Zhou, M.; Zhou, H.; Zhao, D.; Gotor, C.; Romero, L.C.; Shen, J.; Ge, Z.; Zhang, Z.; Shen, W.; et al. Hydrogen sulfide, a signaling molecule in plant stress responses. J. Integr. Plant. Biol. 2021, 63, 146–160. [Google Scholar] [CrossRef]
  9. Chen, H.J.; Ngowi, E.E.; Qian, L.; Li, T.; Qin, Y.Z.; Zhou, J.J.; Li, K.; Ji, X.Y.; Wu, D.D. Role of Hydrogen Sulfide in the Endocrine System. Front. Endocrinol. 2021, 12, 704620. [Google Scholar] [CrossRef]
  10. Krylatov, A.; Maslov, L.; Tsibulnikov, S.Y.; Voronkov, N.; Boshchenko, A.; Downey, J.; Mentzer, R. The Role of Reactive Oxygen Species, Kinases, Hydrogen Sulfide, and Nitric Oxide in the Regulation of Autophagy and Their Impact on Ischemia and Reperfusion Injury in the Heart. Curr. Cardiol. Rev. 2021, 17, e230421186874. [Google Scholar] [CrossRef]
  11. Li, J.; Li, M.; Li, L.; Ma, J.; Yao, C.; Yao, S. Hydrogen sulfide attenuates ferroptosis and stimulates autophagy by blocking mTOR signaling in sepsis-induced acute lung injury. Mol. Immunol. 2022, 141, 318–327. [Google Scholar] [CrossRef] [PubMed]
  12. Huang, S.; Huang, P.; Lin, Z.; Liu, X.; Xu, X.; Guo, L.; Shen, X.; Li, C.; Zhong, Y. Hydrogen sulfide supplement attenuates the apoptosis of retinal ganglion cells in experimental glaucoma. Exp. Eye Res. 2018, 168, 33–48. [Google Scholar] [CrossRef] [PubMed]
  13. Ascenzi, F.; De Vitis, C.; Maugeri-Sacca, M.; Napoli, C.; Ciliberto, G.; Mancini, R. SCD1, autophagy and cancer: Implications for therapy. J. Exp. Clin. Cancer Res. 2021, 40, 265. [Google Scholar] [CrossRef] [PubMed]
  14. Zhao, H.; Liu, H.; Yang, Y.; Wang, H. The Role of Autophagy and Pyroptosis in Liver Disorders. Int. J. Mol. Sci. 2022, 23, 6208. [Google Scholar] [CrossRef]
  15. Grootaert, M.O.J.; Roth, L.; Schrijvers, D.M.; De Meyer, G.R.Y.; Martinet, W. Defective Autophagy in Atherosclerosis: To Die or to Senesce? Oxid. Med. Cell Longev. 2018, 2018, 7687083. [Google Scholar] [CrossRef]
  16. Chen, C.; Lu, L.; Yan, S.; Yi, H.; Yao, H.; Wu, D.; He, G.; Tao, X.; Deng, X. Autophagy and doxorubicin resistance in cancer. Anticancer. Drugs 2018, 29, 1–9. [Google Scholar] [CrossRef]
  17. Wun, C.L.; Quan, Y.; Zhuang, X. Recent Advances in Membrane Shaping for Plant Autophagosome Biogenesis. Front. Plant. Sci. 2020, 11, 565. [Google Scholar] [CrossRef] [PubMed]
  18. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef]
  19. Lu, X.; Ding, Y.; Liu, H.; Sun, M.; Chen, C.; Yang, Y.; Wang, H. The Role of Hydrogen Sulfide Regulation of Autophagy in Liver Disorders. Int. J. Mol. Sci. 2022, 23, 4035. [Google Scholar] [CrossRef]
  20. Lv, S.; Wang, H.; Li, X. The Role of the Interplay between Autophagy and NLRP3 Inflammasome in Metabolic Disorders. Front. Cell Dev. Biol. 2021, 9, 634118. [Google Scholar] [CrossRef]
  21. Lv, S.; Liu, H.; Wang, H. The Interplay between Autophagy and NLRP3 Inflammasome in Ischemia/Reperfusion Injury. Int. J. Mol. Sci. 2021, 22, 8773. [Google Scholar] [CrossRef] [PubMed]
  22. Zhao, S.; Li, X.; Wang, J.; Wang, H. The Role of the Effects of Autophagy on NLRP3 Inflammasome in Inflammatory Nervous System Diseases. Front. Cell Dev. Biol. 2021, 9, 657478. [Google Scholar] [CrossRef]
  23. Zhao, S.; Wang, H. EVA1A Plays an Important Role by Regulating Autophagy in Physiological and Pathological Processes. Int. J. Mol. Sci. 2021, 22, 6181. [Google Scholar] [CrossRef] [PubMed]
  24. Lv, S.; Liu, H.; Wang, H. Exogenous Hydrogen Sulfide Plays an Important Role by Regulating Autophagy in Diabetic-Related Diseases. Int. J. Mol. Sci. 2021, 22, 6715. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, H.; Yang, Y.; Si, X.; Liu, H.; Wang, H. The Role of Pyroptosis and Autophagy in Ischemia Reperfusion Injury. Biomolecules 2022, 12, 1010. [Google Scholar] [CrossRef]
  26. Chen, R.H.; Chen, Y.H.; Huang, T.Y. Ubiquitin-mediated regulation of autophagy. J. Biomed. Sci. 2019, 26, 80. [Google Scholar] [CrossRef]
  27. Wang, J.; Wu, D.; Wang, H. Hydrogen sulfide plays an important protective role by influencing autophagy in diseases. Physiol. Res. 2019, 68, 335–345. [Google Scholar] [CrossRef]
  28. Allaire, M.; Rautou, P.E.; Codogno, P.; Lotersztajn, S. Autophagy in liver diseases: Time for translation? J. Hepatol. 2019, 70, 985–998. [Google Scholar] [CrossRef]
  29. Ploumi, C.; Papandreou, M.E.; Tavernarakis, N. The complex interplay between autophagy and cell death pathways. Biochem. J. 2022, 479, 75–90. [Google Scholar] [CrossRef]
  30. Arif, Y.; Hayat, S.; Yusuf, M.; Bajguz, A. Hydrogen sulfide: A versatile gaseous molecule in plants. Plant. Physiol. Biochem. 2021, 158, 372–384. [Google Scholar] [CrossRef]
  31. Coavoy-Sanchez, S.A.; Costa, S.K.P.; Muscara, M.N. Hydrogen sulfide and dermatological diseases. Br. J. Pharmacol. 2020, 177, 857–865. [Google Scholar] [CrossRef] [PubMed]
  32. Zaorska, E.; Tomasova, L.; Koszelewski, D.; Ostaszewski, R.; Ufnal, M. Hydrogen Sulfide in Pharmacotherapy, beyond the Hydrogen Sulfide-donors. Biomolecules 2020, 10, 323. [Google Scholar] [CrossRef] [PubMed]
  33. Gotor, C.; Garcia, I.; Aroca, A.; Laureano-Marin, A.M.; Arenas-Alfonseca, L.; Jurado-Flores, A.; Moreno, I.; Romero, L.C. Signaling by hydrogen sulfide and cyanide through post-translational modification. J. Exp. Bot. 2019, 70, 4251–4265. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, H.; Bai, Z.; Zhu, L.; Liang, Y.; Fan, X.; Li, J.; Wen, H.; Shi, T.; Zhao, Q.; Wang, Z. Hydrogen sulfide donors: Therapeutic potential in anti-atherosclerosis. Eur. J. Med. Chem. 2020, 205, 112665. [Google Scholar] [CrossRef]
  35. Sokolov, A.S.; Nekrasov, P.V.; Shaposhnikov, M.V.; Moskalev, A.A. Hydrogen sulfide in longevity and pathologies: Inconsistency is malodorous. Ageing Res. Rev. 2021, 67, 101262. [Google Scholar] [CrossRef] [PubMed]
  36. Zhao, H.; Liu, H.; Yang, Y.; Lan, T.; Wang, H.; Wu, D. Hydrogen Sulfide Plays an Important Role by Regulating Endoplasmic Reticulum Stress in Diabetes-Related Diseases. Int. J. Mol. Sci. 2022, 23, 7170. [Google Scholar] [CrossRef] [PubMed]
  37. Lv, S.; Wang, Z.; Wang, J.; Wang, H. Exogenous Hydrogen Sulfide Plays an Important Role Through Regulating Autophagy in Ischemia/Reperfusion Injury. Front. Mol. Biosci. 2021, 8, 681676. [Google Scholar] [CrossRef]
  38. Zhao, S.; Li, X.; Lu, P.; Li, X.; Sun, M.; Wang, H. The Role of the Signaling Pathways Involved in the Effects of Hydrogen Sulfide on Endoplasmic Reticulum Stress. Front. Cell Dev. Biol. 2021, 9, 646723. [Google Scholar] [CrossRef]
  39. Wang, Y.; Yu, R.; Wu, L.; Yang, G. Hydrogen sulfide signaling in regulation of cell behaviors. Nitric Oxide 2020, 103, 9–19. [Google Scholar] [CrossRef]
  40. Khellaf, A.; Khan, D.Z.; Helmy, A. Recent advances in traumatic brain injury. J. Neurol. 2019, 266, 2878–2889. [Google Scholar] [CrossRef] [Green Version]
  41. Khatri, N.; Thakur, M.; Pareek, V.; Kumar, S.; Sharma, S.; Datusalia, A.K. Oxidative Stress: Major Threat in Traumatic Brain Injury. CNS Neurol. Disord. Drug Targets 2018, 17, 689–695. [Google Scholar] [CrossRef] [PubMed]
  42. Najem, D.; Rennie, K.; Ribecco-Lutkiewicz, M.; Ly, D.; Haukenfrers, J.; Liu, Q.; Nzau, M.; Fraser, D.D.; Bani-Yaghoub, M. Traumatic brain injury: Classification, models, and markers. Biochem. Cell Biol. 2018, 96, 391–406. [Google Scholar] [CrossRef] [PubMed]
  43. Gao, C.; Yan, Y.; Chen, G.; Wang, T.; Luo, C.; Zhang, M.; Chen, X.; Tao, L. Autophagy Activation Represses Pyroptosis through the IL-13 and JAK1/STAT1 Pathways in a Mouse Model of Moderate Traumatic Brain Injury. ACS Chem. Neurosci. 2020, 11, 4231–4239. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, J.Y.; Lee, J.H.; Gu, X.; Wei, Z.Z.; Harris, M.J.; Yu, S.P.; Wei, L. Intranasally Delivered Wnt3a Improves Functional Recovery after Traumatic Brain Injury by Modulating Autophagic, Apoptotic, and Regenerative Pathways in the Mouse Brain. J. Neurotrauma 2018, 35, 802–813. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, M.; Shan, H.; Chang, P.; Wang, T.; Dong, W.; Chen, X.; Tao, L. Hydrogen sulfide offers neuroprotection on traumatic brain injury in parallel with reduced apoptosis and autophagy in mice. PLoS ONE 2014, 9, e87241. [Google Scholar] [CrossRef] [PubMed]
  46. Tian, J.; Yang, L.; Wang, P.; Yang, L.; Fan, Z. Exogenous CGRP Regulates Apoptosis and Autophagy to Alleviate Traumatic Brain Injury Through Akt/mTOR Signalling Pathway. Neurochem. Res. 2020, 45, 2926–2938. [Google Scholar] [CrossRef]
  47. Oberstein, A.; Jeffrey, P.D.; Shi, Y. Crystal structure of the Bcl-XL-Beclin 1 peptide complex: Beclin 1 is a novel BH3-only protein. J. Biol. Chem. 2007, 282, 13123–13132. [Google Scholar] [CrossRef]
  48. Teresak, P.; Lapao, A.; Subic, N.; Boya, P.; Elazar, Z.; Simonsen, A. Regulation of PRKN-independent mitophagy. Autophagy 2022, 18, 24–39. [Google Scholar] [CrossRef]
  49. Scatena, R. Mitochondria and drugs. Adv. Exp. Med. Biol. 2012, 942, 329–346. [Google Scholar]
  50. Ahluwalia, M.; Kumar, M.; Ahluwalia, P.; Rahimi, S.; Vender, J.R.; Raju, R.P.; Hess, D.C.; Baban, B.; Vale, F.L.; Dhandapani, K.M.; et al. Rescuing mitochondria in traumatic brain injury and intracerebral hemorrhages—A potential therapeutic approach. Neurochem. Int. 2021, 150, 105192. [Google Scholar] [CrossRef]
  51. Palzur, E.; Edelman, D.; Sakas, R.; Soustiel, J.F. Etifoxine Restores Mitochondrial Oxidative Phosphorylation and Improves Cognitive Recovery Following Traumatic Brain Injury. Int. J. Mol. Sci. 2021, 22, 12881. [Google Scholar] [CrossRef] [PubMed]
  52. Zhao, X.J.; Zhu, H.Y.; Wang, X.L.; Lu, X.W.; Pan, C.L.; Xu, L.; Liu, X.; Xu, N.; Zhang, Z.Y. Oridonin Ameliorates Traumatic Brain Injury-Induced Neurological Damage by Improving Mitochondrial Function and Antioxidant Capacity and Suppressing Neuroinflammation through the Nrf2 Pathway. J. Neurotrauma 2022, 39, 530–543. [Google Scholar] [CrossRef] [PubMed]
  53. Xu, K.; Wu, F.; Xu, K.; Li, Z.; Wei, X.; Lu, Q.; Jiang, T.; Wu, F.; Xu, X.; Xiao, J.; et al. NaHS restores mitochondrial function and inhibits autophagy by activating the PI3K/Akt/mTOR signalling pathway to improve functional recovery after traumatic brain injury. Chem. Biol. Interact. 2018, 286, 96–105. [Google Scholar] [CrossRef] [PubMed]
  54. Li, M.; Xu, C.; Shi, J.; Ding, J.; Wan, X.; Chen, D.; Gao, J.; Li, C.; Zhang, J.; Lin, Y.; et al. Fatty acids promote fatty liver disease via the dysregulation of 3-mercaptopyruvate sulfurtransferase/hydrogen sulfide pathway. Gut 2018, 67, 2169–2180. [Google Scholar] [CrossRef]
  55. Zhang, F.; Chen, S.; Wen, J.Y.; Chen, Z.W. 3-Mercaptopyruvate sulfurtransferase/hydrogen sulfide protects cerebral endothelial cells against oxygen-glucose deprivation/reoxygenation-induced injury via mitoprotection and inhibition of the RhoA/ROCK pathway. Am. J. Physiol. Cell Physiol. 2020, 319, C720–C733. [Google Scholar] [CrossRef]
  56. Zhang, M.; Shan, H.; Chang, P.; Ma, L.; Chu, Y.; Shen, X.; Wu, Q.; Wang, Z.; Luo, C.; Wang, T.; et al. Upregulation of 3-MST Relates to Neuronal Autophagy after Traumatic Brain Injury in Mice. Cell Mol. Neurobiol. 2017, 37, 291–302. [Google Scholar] [CrossRef]
  57. Ahsan, A.; Liu, M.; Zheng, Y.; Yan, W.; Pan, L.; Li, Y.; Ma, S.; Zhang, X.; Cao, M.; Wu, Z.; et al. Natural compounds modulate the autophagy with potential implication of stroke. Acta Pharm. Sin. B 2021, 11, 1708–1720. [Google Scholar] [CrossRef]
  58. Wu, X.; Zheng, Y.; Liu, M.; Li, Y.; Ma, S.; Tang, W.; Yan, W.; Cao, M.; Zheng, W.; Jiang, L.; et al. BNIP3L/NIX degradation leads to mitophagy deficiency in ischemic brains. Autophagy 2021, 17, 1934–1946. [Google Scholar] [CrossRef]
  59. Zheng, W.; Liu, B.; Shi, E. Perillaldehyde Alleviates Spinal Cord Ischemia-Reperfusion Injury Via Activating the Nrf2 Pathway. J. Surg. Res. 2021, 268, 308–317. [Google Scholar] [CrossRef]
  60. Li, H.; Dong, X.; Yang, Y.; Jin, M.; Cheng, W. The Neuroprotective Mechanism of Spinal Cord Stimulation in Spinal Cord Ischemia/Reperfusion Injury. Neuromodulation 2021, 24, 43–48. [Google Scholar] [CrossRef]
  61. Jing, W.; Zhang, T.; Jiang, W.; Zhang, T. Neuroprotective effect of neuregulin-1beta on spinal cord ischemia reperfusion injury. J. Spinal Cord Med. 2021, 44, 583–589. [Google Scholar] [CrossRef] [PubMed]
  62. Yin, J.; Yin, Z.; Wang, B.; Zhu, C.; Sun, C.; Liu, X.; Gong, G. Angiopoietin-1 Protects Spinal Cord Ischemia and Reperfusion Injury by Inhibiting Autophagy in Rats. Neurochem. Res. 2019, 44, 2746–2754. [Google Scholar] [CrossRef] [PubMed]
  63. Gu, C.; Li, L.; Huang, Y.; Qian, D.; Liu, W.; Zhang, C.; Luo, Y.; Zhou, Z.; Kong, F.; Zhao, X.; et al. Salidroside Ameliorates Mitochondria-Dependent Neuronal Apoptosis after Spinal Cord Ischemia-Reperfusion Injury Partially through Inhibiting Oxidative Stress and Promoting Mitophagy. Oxid. Med. Cell Longev. 2020, 2020, 3549704. [Google Scholar] [CrossRef]
  64. Li, L.; Jiang, H.K.; Li, Y.P.; Guo, Y.P. Hydrogen sulfide protects spinal cord and induces autophagy via miR-30c in a rat model of spinal cord ischemia-reperfusion injury. J. Biomed. Sci. 2015, 22, 50. [Google Scholar] [CrossRef] [PubMed]
  65. Yang, X.; Zhong, X.; Tanyi, J.L.; Shen, J.; Xu, C.; Gao, P.; Zheng, T.M.; DeMichele, A.; Zhang, L. mir-30d Regulates multiple genes in the autophagy pathway and impairs autophagy process in human cancer cells. Biochem. Biophys. Res. Commun. 2013, 431, 617–622. [Google Scholar] [CrossRef] [PubMed]
  66. Hyrskyluoto, A.; Reijonen, S.; Kivinen, J.; Lindholm, D.; Korhonen, L. GADD34 mediates cytoprotective autophagy in mutant huntingtin expressing cells via the mTOR pathway. Exp. Cell Res. 2012, 318, 33–42. [Google Scholar] [CrossRef] [PubMed]
  67. Novak, C.M.; Ozen, M.; Burd, I. Perinatal Brain Injury: Mechanisms, Prevention, and Outcomes. Clin. Perinatol. 2018, 45, 357–375. [Google Scholar] [CrossRef]
  68. Arteaga, O.; Alvarez, A.; Revuelta, M.; Santaolalla, F.; Urtasun, A.; Hilario, E. Role of Antioxidants in Neonatal Hypoxic-Ischemic Brain Injury: New Therapeutic Approaches. Int. J. Mol. Sci. 2017, 18, 265. [Google Scholar] [CrossRef]
  69. Zhang, X.; Bian, J.S. Hydrogen sulfide: A neuromodulator and neuroprotectant in the central nervous system. ACS Chem. Neurosci. 2014, 5, 876–883. [Google Scholar] [CrossRef]
  70. Zhong, H.; Yu, H.; Chen, J.; Sun, J.; Guo, L.; Huang, P.; Zhong, Y. Hydrogen Sulfide and Endoplasmic Reticulum Stress: A Potential Therapeutic Target for Central Nervous System Degeneration Diseases. Front. Pharmacol. 2020, 11, 702. [Google Scholar] [CrossRef]
  71. Xin, D.; Chu, X.; Bai, X.; Ma, W.; Yuan, H.; Qiu, J.; Liu, C.; Li, T.; Zhou, X.; Chen, W.; et al. l-Cysteine suppresses hypoxia-ischemia injury in neonatal mice by reducing glial activation, promoting autophagic flux and mediating synaptic modification via H2S formation. Brain Behav. Immun. 2018, 73, 222–234. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, H.; Zhong, P.; Sun, L. Exogenous hydrogen sulfide mitigates NLRP3 inflammasome-mediated inflammation through promoting autophagy via the AMPK-mTOR pathway. Biol. Open 2019, 8, bio043653. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, X.; Li, H.; Gong, Y.; Zheng, H.; Zhao, D. Hydrogen sulfide ameliorated lipopolysaccharide-induced acute lung injury by inhibiting autophagy through PI3K/Akt/mTOR pathway in mice. Biochem. Biophys. Res. Commun. 2018, 507, 514–518. [Google Scholar] [CrossRef]
  74. You, L.; Wang, Z.; Li, H.; Shou, J.; Jing, Z.; Xie, J.; Sui, X.; Pan, H.; Han, W. The role of STAT3 in autophagy. Autophagy 2015, 11, 729–739. [Google Scholar] [CrossRef] [PubMed]
  75. Cherubini, J.M.; Cheng, J.L.; Williams, J.S.; MacDonald, M.J. Sleep deprivation and endothelial function: Reconciling seminal evidence with recent perspectives. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H29–H35. [Google Scholar] [CrossRef] [PubMed]
  76. Olaithe, M.; Bucks, R.S.; Hillman, D.R.; Eastwood, P.R. Cognitive deficits in obstructive sleep apnea: Insights from a meta-review and comparison with deficits observed in COPD, insomnia, and sleep deprivation. Sleep Med. Rev. 2018, 38, 39–49. [Google Scholar] [CrossRef] [PubMed]
  77. Qiu, C.; Wang, M.; Yu, W.; Rong, Z.; Zheng, H.S.; Sun, T.; Liu, S.B.; Zhao, M.G.; Wu, Y.M. Activation of the Hippocampal LXRbeta Improves Sleep-Deprived Cognitive Impairment by Inhibiting Neuroinflammation. Mol. Neurobiol. 2021, 58, 5272–5288. [Google Scholar] [CrossRef]
  78. Duan, H.; Li, L.; Shen, S.; Ma, Y.; Yin, X.; Liu, Z.; Yuan, C.; Wang, Y.; Zhang, J. Hydrogen Sulfide Reduces Cognitive Impairment in Rats after Subarachnoid Hemorrhage by Ameliorating Neuroinflammation Mediated by the TLR4/NF-kappaB Pathway in Microglia. Front. Cell Neurosci. 2020, 14, 210. [Google Scholar] [CrossRef]
  79. Gao, S.; Tang, Y.Y.; Jiang, L.; Lan, F.; Li, X.; Zhang, P.; Zou, W.; Chen, Y.J.; Tang, X.Q. H2S Attenuates Sleep Deprivation-Induced Cognitive Impairment by Reducing Excessive Autophagy via Hippocampal Sirt-1 in WISTAR RATS. Neurochem. Res. 2021, 46, 1941–1952. [Google Scholar] [CrossRef]
  80. Zuo, J.X.; Li, M.; Jiang, L.; Lan, F.; Tang, Y.Y.; Kang, X.; Zou, W.; Wang, C.Y.; Zhang, P.; Tang, X.Q. Hydrogen Sulfide Prevents Sleep Deprivation-Induced Hippocampal Damage by Upregulation of Sirt1 in the Hippocampus. Front. Neurosci. 2020, 14, 169. [Google Scholar] [CrossRef]
  81. Yang, S.Q.; Jiang, L.; Lan, F.; Wei, H.J.; Xie, M.; Zou, W.; Zhang, P.; Wang, C.Y.; Xie, Y.R.; Tang, X.Q. Inhibited Endogenous H2S Generation and Excessive Autophagy in Hippocampus Contribute to Sleep Deprivation-Induced Cognitive Impairment. Front. Psychol. 2019, 10, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Sartorius, N. Depression and diabetes. Dialogues Clin. Neurosci. 2018, 20, 47–52. [Google Scholar] [CrossRef]
  83. Zhang, Y.; Chen, Y.; Ma, L. Depression and cardiovascular disease in elderly: Current understanding. J. Clin. Neurosci. 2018, 47, 1–5. [Google Scholar] [CrossRef] [PubMed]
  84. Bruijniks, S.J.E.; van Grootheest, G.; Cuijpers, P.; de Kluiver, H.; Vinkers, C.H.; Peeters, F.; Penninx, B.; Teunissen, C.E.; Huibers, M.J.H. Working memory moderates the relation between the brain-derived neurotropic factor (BDNF) and psychotherapy outcome for depression. J. Psychiatr. Res. 2020, 130, 424–432. [Google Scholar] [CrossRef] [PubMed]
  85. Kamdi, S.P.; Raval, A.; Nakhate, K.T. Phloridzin ameliorates type 2 diabetes-induced depression in mice by mitigating oxidative stress and modulating brain-derived neurotrophic factor. J. Diabetes Metab. Disord. 2021, 20, 341–348. [Google Scholar] [CrossRef]
  86. Liu, H.Y.; Wei, H.J.; Wu, L.; Liu, S.M.; Tang, Y.Y.; Zou, W.; Wang, C.Y.; Zhang, P.; Tang, X.Q. BDNF-TrkB pathway mediates antidepressant-like roles of H2 S in diabetic rats via promoting hippocampal autophagy. Clin. Exp. Pharmacol. Physiol. 2020, 47, 302–312. [Google Scholar] [CrossRef]
  87. Liu, J.; Wang, Y.H.; Li, W.; Liu, L.; Yang, H.; Meng, P.; Han, Y.S. Structural and functional damage to the hippocampal neurovascular unit in diabetes-related depression. Neural Regen. Res. 2019, 14, 289–297. [Google Scholar]
  88. Zhou, X.Y.; Zhang, F.; Hu, X.T.; Chen, J.; Tang, R.X.; Zheng, K.Y.; Song, Y.J. Depression can be prevented by astaxanthin through inhibition of hippocampal inflammation in diabetic mice. Brain Res. 2017, 1657, 262–268. [Google Scholar] [CrossRef]
  89. Bidesi, N.S.R.; Vang Andersen, I.; Windhorst, A.D.; Shalgunov, V.; Herth, M.M. The role of neuroimaging in Parkinson’s disease. J. Neurochem. 2021, 159, 660–689. [Google Scholar] [CrossRef]
  90. Waller, S.; Williams, L.; Morales-Briceno, H.; Fung, V.S. The initial diagnosis and management of Parkinson’s disease. Aust. J. Gen. Pract. 2021, 50, 793–800. [Google Scholar] [CrossRef]
  91. Kumar, S.; Behl, T.; Sehgal, A.; Singh, S.; Sharma, N.; Bhatia, S.; Al-Harassi, A.; Abdel-Daim, M.M.; Bungau, S. Exploring the Role of Orexinergic Neurons in Parkinson’s Disease. Neurotox. Res. 2021, 39, 2141–2153. [Google Scholar] [CrossRef] [PubMed]
  92. Zhao, Y.; Zhang, Z.; Qin, S.; Fan, W.; Li, W.; Liu, J.; Wang, S.; Xu, Z.; Zhao, M. Acupuncture for Parkinson’s Disease: Efficacy Evaluation and Mechanisms in the Dopaminergic Neural Circuit. Neural Plast. 2021, 2021, 9926445. [Google Scholar] [CrossRef] [PubMed]
  93. Gordon, R.; Albornoz, E.A.; Christie, D.C.; Langley, M.R.; Kumar, V.; Mantovani, S.; Robertson, A.A.B.; Butler, M.S.; Rowe, D.B.; O’Neill, L.A.; et al. Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice. Sci. Transl. Med. 2018, 10, eaah4066. [Google Scholar] [CrossRef] [PubMed]
  94. Jiang, W.; Zou, W.; Hu, M.; Tian, Q.; Xiao, F.; Li, M.; Zhang, P.; Chen, Y.J.; Jiang, J.M. Hydrogen sulphide attenuates neuronal apoptosis of substantia nigra by re-establishing autophagic flux via promoting leptin signalling in a 6-hydroxydopamine rat model of Parkinson’s disease. Clin. Exp. Pharmacol. Physiol. 2022, 49, 122–133. [Google Scholar] [CrossRef] [PubMed]
  95. Huang, M.; Lou, D.; Charli, A.; Kong, D.; Jin, H.; Zenitsky, G.; Anantharam, V.; Kanthasamy, A.; Wang, Z.; Kanthasamy, A.G. Mitochondrial dysfunction-induced H3K27 hyperacetylation perturbs enhancers in Parkinson’s disease. JCI Insight 2021, 6, e138088. [Google Scholar] [CrossRef] [PubMed]
  96. Ahn, E.H.; Lei, K.; Kang, S.S.; Wang, Z.H.; Liu, X.; Hong, W.; Wang, Y.T.; Edgington-Mitchell, L.E.; Jin, L.; Ye, K. Mitochondrial dysfunction triggers the pathogenesis of Parkinson’s disease in neuronal C/EBPbeta transgenic mice. Mol. Psychiatry 2021, 26, 7838–7850. [Google Scholar] [CrossRef]
  97. Jiao, D.; Yang, Z.; Wang, L.; Hu, B.; Wang, J.; Xu, A.; Cheng, W.; Jia, B.; Qing, Y.; Zhao, H.Y.; et al. Endogenous leptin promotes autophagy in EBSS-induced PFCs. Anim. Cells Syst. 2019, 23, 318–325. [Google Scholar] [CrossRef]
  98. Zhang, W.; Li, Z.L.; Zhang, Q. Leptin inhibits apoptosis of nucleus pulposus cells via promoting autophagy. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4065. [Google Scholar]
  99. Wu, D.; Zhong, P.; Wang, J.; Wang, H. Exogenous hydrogen sulfide mitigates LPS + ATP-induced inflammation by inhibiting NLRP3 inflammasome activation and promoting autophagy in L02 cells. Mol. Cell Biochem. 2019, 457, 145–156. [Google Scholar] [CrossRef]
Figure 1. Diagram of three types of autophagy processes.
Figure 1. Diagram of three types of autophagy processes.
Metabolites 12 00879 g001
Figure 2. Diagram of the endogenous H2S generation process.
Figure 2. Diagram of the endogenous H2S generation process.
Metabolites 12 00879 g002
Table 1. The summary of the role of H2S targeting autophagy in the pathological processes of the nervous system.
Table 1. The summary of the role of H2S targeting autophagy in the pathological processes of the nervous system.
The Type of Nervous System DiseasesThe Role of H2S Targeting AutophagyExperimental ModelReference
Traumatic brain injury (TBI)Exogenous H2S ameliorates TBI of mice through suppressing autophagy and apoptosisMice model of TBI[45]
TBIExogenous H2S improves TBI by inhibiting autophagy via activating PI3K/AKT/mTOR pathwayMice/SH-SY5Y cells model of TBI[53]
TBIEndogenous H2S may play protective role against TBI by inhibiting autophagic cell deathMice model of TBI[56]
Spinal cord ischemia-reperfusion (I/R) injuryExogenous H2S ameliorates spinal I/R injury through promoting autophagy and clearing the damaged mitochondria caused by I/R injuryRat model of spinal cord ischemia-reperfusion injury[64]
hypoxia-ischemia (HI) brain injuryEndogenous H2S produced by L-Cysteine improves HI-induced brain injury of neonatal mice by promoting autophagy via inhibiting the mTOR and Stat3 pathwayNeonatal mice model of hypoxia-ischemia injury[71]
sleep deprivation (SD)-induced cognitive impairmentExogenous H2S alleviates SD-induced cognitive impairment by inhibiting autophagy via hippocampal Sirt-1Mice model of SD-induced cognitive impairment[79]
diabetes depressionExogenous H2S ameliorates depression through promoting autophagy by activating BDNF-TrkB pathwayRat model of diabetes depression[86]
Parkinson’s disease (PD)Exogenous H2S improves neuronal apoptosis in substantia nigra through promoting autophagy impaired by 6-OHDA via activating leptin signaling6-hydroxydopamine rat model of PD[94]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhao, H.; Yang, Y.; Liu, H.; Wang, H. The Role of Hydrogen Sulfide Targeting Autophagy in the Pathological Processes of the Nervous System. Metabolites 2022, 12, 879. https://doi.org/10.3390/metabo12090879

AMA Style

Zhao H, Yang Y, Liu H, Wang H. The Role of Hydrogen Sulfide Targeting Autophagy in the Pathological Processes of the Nervous System. Metabolites. 2022; 12(9):879. https://doi.org/10.3390/metabo12090879

Chicago/Turabian Style

Zhao, Huijie, Yihan Yang, Huiyang Liu, and Honggang Wang. 2022. "The Role of Hydrogen Sulfide Targeting Autophagy in the Pathological Processes of the Nervous System" Metabolites 12, no. 9: 879. https://doi.org/10.3390/metabo12090879

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop